Next Article in Journal
Essential Oil Fumigation Modulates Nutrient Content in Selected Mushrooms During Postharvest Storage
Previous Article in Journal
Applications of the Cellular Thermal Shift Assay to Drug Discovery in Natural Products: A Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Drug Resistance Analysis of Pancreatic Cancer Based on Universally Differentially Expressed Genes

1
School of Biology and Engineering, Guizhou Medical University, Guiyang 550025, China
2
Fujian Key Laboratory of Medical Bioinformatics, Department of Bioinformatics, School of Medical Technology and Engineering, Fujian Medical University, Fuzhou 350100, China
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(9), 3936; https://doi.org/10.3390/ijms26093936
Submission received: 14 February 2025 / Revised: 10 April 2025 / Accepted: 11 April 2025 / Published: 22 April 2025
(This article belongs to the Section Molecular Genetics and Genomics)

Abstract

:
The high heterogeneity between patients can complicate the diagnosis and treatment of pancreatic ductal adenocarcinoma (PDAC). Here, we explored the association of universally differentially expressed genes (UDEGs) with resistance to chemotherapy and immunotherapy in the context of pancreatic cancer. In this work, sixteen up-regulated and three down-regulated genes that were dysregulated in more than 85% of 102 paired and 5% of 521 unpaired PDAC samples were identified and defined as UDEGs. A single-cell level analysis further validated the high expression levels of the up-UDEGs and the low levels of the down-UDEGs in cancer-related ductal cells, which could represent the malignant changes seen in pancreatic cancer. Based on a drug sensitivity analysis, we found that ANLN, GPRC5A and SERPINB5 are closely related to the resistance mechanism of PDAC, and their high expression predicted worse survival for PDAC patients. This suggests that targeting these genes could be a potential way to reduce drug resistance and improve survival. Based on the immune infiltration analysis, the abnormal expression of the UDEGs was found to be related to the formation of an immunosuppressive tumor microenvironment. In conclusion, these UDEGs are common features of PDAC and could be involved in the resistance of pancreatic cancer and might serve as novel drug targets to guide research into drug repurposing.

1. Introduction

Pancreatic ductal adenocarcinoma (PDAC) is considered one of the most aggressive malignant neoplasms, with a 12.8% five-year survival rate according to National Cancer Institute statistics. Complete surgery resection is the only possible cure for resectable PDAC, but most patients are already in an advanced stage at diagnosis [1]. Chemotherapy with/without radiotherapy has been a mainstay strategy for treating PDAC, but it only provides a modest improvement in survival [2]. Only 3% to 11% of patients have pathological complete responses after receiving chemotherapy with/without radiation therapy [3,4,5,6,7], which may be due to the high heterogeneity between patients and the ubiquitous drug resistance of PDAC. Immune checkpoint blockade (ICB) has changed the standard of care for multiple cancers and has brought hope to pancreatic cancer patients; however, almost all tentative trials have had little effect or failed before clinical application [8,9]. The most common reason for failure was the immunosuppressive tumor microenvironment (TME) resulting in low effector T cell infiltration, which is unfavorable for the immune response [10]. We suspect that there are some common features underlying the high heterogeneity of pancreatic cancer, which could be closely related to the mechanism underlying the ineffectiveness of current therapies. It is essential to identify and target these mechanisms to improve the early diagnosis rate and therapeutic efficiency.
Conventional differentially expressed gene (DEG) screening methods for bulk tissues only compare the average expression levels between disease and control groups, which cannot show the dysregulation status of each gene in a single disease sample or the frequency of differential expression of genes in all disease samples. The gold standard for individualized DEG screening is comparing the expression levels between paired tumor and adjacent normal samples from the same patient. However, the late diagnosis and difficulty of sampling pathologically normal samples in the complicated structure of the pancreas has led to few paired samples. To solve this problem, a robust individual DEG screening method, RankComp [11], has been developed to detect the dysregulation of genes in a single tumor sample relative to the normal background derived from normal samples from different studies. Thus, we can identify individual-level DEGs for unpaired tumor samples based on the aggregation of normal samples using this method. Here, we made full use of paired and unpaired PDAC samples to identify common molecular characteristics of PDAC patients to construct a robust early diagnosis model and determine the mechanisms underlying the resistance to current therapies.
In this study, using transcription profiles of paired and unpaired PDAC bulk samples, we identified individual-level DEGs in the PDAC samples and defined the genes that were dysregulated in at least 85% of the PDAC samples as universally differentially expressed genes (UDEGs). Bulk assays represent a population average, which masks the heterogeneity that exists at the single-cell level [12,13]. Thus, we analyzed the expression of the UDEGs at the single-cell level to explore their roles in the formation of tumors. Finally, we investigated the potential association between the UDEGs and overall drug resistance based on GDSC resistance data, as well as their correlation with responsiveness to immunotherapy using reported immunotherapy biomarkers.

2. Results

2.1. Identification of UDEGs in PDAC

The workflow for identifying UDEGs is shown in Figure 1A. In the 102 paired cancer and normal samples from GSE60646, GSE22780, GSE28735 and GSE15471, 180 genes were up-regulated and 32 genes were down-regulated in more than 85% of the tumor samples compared to the paired adjacent normal samples (Figure 1B). In the unpaired samples, based on 44,157,295 gene pairs that were stable in at least 90% of the 212 normal samples, DEGs were detected for each tumor sample using RankComp (FDR < 0.05). A total of 207 up-regulated and 153 down-regulated genes with dysregulated frequencies greater than 85% in 521 PDAC samples were detected. Finally, sixteen up-regulated and three down-regulated genes overlapped in the two DEG lists and were defined as UDEGs (Figure 1B–D).

2.2. Functions and Roles of UDEGs in PDAC

The P53 signaling pathway (p = 2.76 × 10−3) and cancer gene set from the CancerMine database (Hypergeometric test, p = 1.45 × 10−3) were enriched with UDEGs. Among them, SFN, LAMC2, GPRC5A, ANLN, ESM1, SERPINB5 and SLC6A14 have been reported to be closely related to the carcinogenesis and metastasis of PDAC (Appendix Table A1). SFN (Stratifin) is over-expressed and hypomethylated in cancer tissues compared with normal ducts and plays a role as an oncogene in pancreatic cancer [14,15]. LAMC2 (laminin subunit gamma 2) promotes cell proliferation, invasion and migration in multiple cancers, including pancreatic cancer [16,17,18]. Knockout of GPRC5A (G protein-coupled receptor family C, group 5, member A) reduced the proliferation and migration ability of pancreatic cancer cell lines and suppressed the resistance of pancreatic cancer cell lines to the chemotherapy drugs gemcitabine, oxaliplatin and fluorouracil [19]. The up-regulation of anillin (ANLN) promotes the progression and metastasis of PDAC [20,21,22]. ESM1 encodes a secreted protein that is mainly expressed in endothelial cells; it regulates the proliferation, migration, invasion and drug resistance of tumor cells, thereby promoting tumor progression and metastasis and playing an important role in the pathogenesis of pancreatic cancer [23]. Several reports have shown that the over-expression of SERPINB5 (serine protease inhibitor B5) can promote PDAC metastasis [24], and the marked up-regulation of SLC6A14 (solute carrier, family 6, member 14) in pancreatic cancer leads to worse survival [25,26,27]. Overall, these UDEGs are important in the carcinogenesis, invasion and metastasis of PDAC.
To further analyze the biological functions of the UDEGs, we generated a PPI network for the 19 UDEGs and found 155 proteins that interact with them (Figure 2A). Several carcinogenesis- and metastasis-related pathways, such as the cell cycle, focal adhesion, ErbB signaling and PI3K-Akt signaling pathways, are enriched with these 174 genes (Figure 2B). According to the Reactome pathway analysis, immune system, signal transduction, cell cycle, gene expression, disease, programmed cell death and extracellular matrix pathways were hierarchically enriched with the UDEGs (Figure 2C). These results indicate that in addition to cell proliferation and adhesion, changes in the immune system are also important for the carcinogenesis and development of pancreatic cancer.

2.3. Characteristics of UDEGs at Single-Cell Level

Because a bulk cancer tissue sample is a mixture of different types of cells [12,13], we explored whether the UDEGs are specifically dysregulated in cancer cells at the single-cell level. Here, we analyzed scRNA-seq profiles from CRA001160, which contains 24 PDAC and 11 control samples. From the tumor samples, 42,066 cells were acquired after quality control, which were separated into 39 clusters (Figure 3A), classified as acinar cells (cluster 20), B cells (clusters 7, 22 and 25), macrophages (clusters 1, 15, 32 and 37), endocrine cells (cluster 31), neuroendocrine cells (cluster 36), fibroblasts (clusters 5, 6, 23, 24, 33 and 38), endothelial cells (clusters 0 and 11), monocytes, stellate cells (clusters 2 and 4) and T cells. In particular, clusters 9, 10, 12, 13, 14, 16, 18, 19, 21, 26, 27, 28, 30, 34 and 35 not only expressed ductal cell markers (KRT19, MMP7, TSPAN8, SOX9 and LCN2), but also expressed cancer cell markers (MUC1 and PROM1). Thus, we defined these clusters as cancer-related ductal cells, and cluster 3, which only expressed ductal cell markers, was defined as ductal cells with normal characteristics.
From the 11 control samples, 15,553 cells passed the quality control and were classified into 27 clusters (Figure 3B), including acinar cells (clusters 7 and 15), ductal cells (clusters 0, 2, 4, 5, 6, 9, 11, 14, 16, 19 and 26), endocrine cells (cluster 20), endothelial cells (clusters 1, 8, 10, 17, 18 and 21), fibroblasts (cluster 3), stellate cells (cluster 12) and a small number of immune cells (T cells (clusters 22 and 24), monocytes (cluster 25), B cells (cluster 23) and macrophages (cluster 13)).
The major distinctions between the tumor and control samples were the large amount (27%) of cancer-related ductal cells and large proportion of immune cells in the tumor samples (25.29% in tumor samples compared with 4.15% in control samples) (Figure 3C,D). Although these cells are from merging different samples, the result still reflects the appearance of malignant lesions and an increase in inflammation in pancreatic cancer. In particular, up-UDEGs were specifically and highly expressed in cancer-related ductal cells while down-UDEGs had low expression (Figure 3E,F). Conversely, the down-UDEGs were expressed at high levels in the control samples, especially in some cell types, such as ductal cells, endothelial cells and fibroblasts. The KEGG enrichment analysis of specific highly expressed genes in each cell type found that most cells exhibit similar functions in both tumor and normal samples, such as T cells, macrophages or endocrine cells. However, multiple cancer-related pathways, such as necroptosis, pancreatic cancer, chemical carcinogenesis and reactive oxygen pathways, and metabolic pathways associated with proliferation, glutathione metabolism and glycolysis/gluconeogenesis (Table 1) were enriched with the DEGs from cancer-related ductal cells, similar to the enrichment in bulk tissues. This result indicates that cancer-related ductal cells contribute most to the markedly high expression levels of up-UDEGs in the tumor tissues compared to the control samples and to the cancer-related functions in PDAC tissues.

2.4. UDEGs Involved in Resistance to Anticancer Drugs

Drug resistance is an important cause of poor chemotherapy responses in pancreatic cancer. We posited that the UDEGs we identified are involved in the universal drug resistance of PDAC patients. We downloaded the expression profiles of 32 untreated pancreatic cell lines and IC50 values of the 448 anticancer drugs used to treat the 32 pancreas cancer cell lines from the GDSC database. The 412 drugs that were used to treat more than half of these cell lines were retained. The mRNA expression profiles of cell lines were measured using the Affymetrix Human Genome U219 Array, which only contains nine of the nineteen UDEGs. Therefore, we only analyzed the relationship between these nine genes and drug resistance in our study. Based on the Spearman correlation analysis of the expression of the nine UDEGs and the IC50 for pancreatic cancer cell lines, 371 drug–gene relationships were found to be significantly correlated (Figure 4A, Appendix Table A2).
Interestingly, most of the relationships between drug resistance and the expression levels of the up-UDEGs ANLN, GPRC5A and SERPINB5 were positive (Figure 4B), indicating that these three genes are closely related to the resistance mechanisms of pancreatic cancer. Moreover, we found that high expression levels of ANLN, GPRC5A and SERPINB5 were significantly associated with poor OS (overall survival) for TCGA PDAC patients (Figure 4C; ANLN: p = 0.034; GPRC5A: p = 0.045; SERPINB5: p = 0.028; log-rank test). Combining the above results, the universal drug resistance of patients with high expression of ANLN, GPRC5A and SERPINB5 might be one of the causes of poor survival. Thus, targeting these genes could be an effective way to suppress drug resistance and improve survival.
Among the significant correlations between LRP8 expression and the IC50s of anticancer compounds, 43/50 (86%) were negative (Figure 4B), indicating that most anticancer drugs exhibit higher efficacy when LRP8 is highly expressed; these drugs included tamoxifen (Rho = −0.49, p = 4.83 × 10−3), rapamycin (Rho = −0.51, p = 3.58 × 10−3) and afuresertib (Rho = −0.50, p = 5.39 × 10−3). Tamoxifen is widely used in the treatment of estrogen receptor-positive breast cancer and has been found to inhibit the myofibroblastic differentiation of pancreatic stellate cells (PSCs) and cell invasion into the tumor microenvironment [28], but it has not been used in the treatment of pancreatic cancer. Rapamycin is a lipophilic macrolide antibiotic that was originally developed as a fungicide and an immunosuppressant. Combined with cisplatin, it can affect the PI3K/AKT/mTOR signal transduction pathway, which leads to markedly increased cell apoptosis, indicating that rapamycin can mediate the sensitivity of pancreatic cancer cells to cisplatin [29]. Afuresertib is a novel inhibitor of the serine/threonine kinase AKT and has shown clinical efficacy as a monotherapy against hematological malignancies and could be used in combination with standard therapies for multiple myeloma [30]. In summary, our results identified some potential novel drugs for pancreatic cancer therapy that deserve further clinical research.
We also explored drug repurposing based on the UDEGs by generating a DGI network for the UDEGs and their PPI partners (Figure 4D). Some anticancer drugs (amatuximab, AZD-4877, ispinesib, letrozole, cergutuzumab amunaleukin, filanesib and nutlin-3), type II diabetes drugs (omega-3-carboxylic acids and ruboxistaurin) and drugs used to stimulate gastric and pancreatic secretions in hyperlipoproteinemia (levocarnitine) were found to interact with the UDEGs. Moreover, some novel drugs that have not been used to treat PDAC-related diseases were found to interact with the UDEGs, such as a drug for ocular hypertension (bimatoprost) and the antiepileptic drug losigamone. These results not only indicate that the UDEGs could be drug targets for pancreatic cancer but also provide new insights for drug repurposing to treat pancreatic cancer.

2.5. Roles of UDEGs in Response to Immunotherapy for PDAC Patients

The challenge in treating pancreatic cancer lies in its resistance to both chemotherapy and radiotherapy, as well as its unresponsiveness to the increasingly popular immunotherapy approach. Thus, we explored the roles of the UDEGs in the immune response to immunotherapy in PDAC patients.
A commonly used biomarker of immunotherapy, TMB, is universally low in PDAC patients, with an average of 0.73 mutations/megabase. For comparison, cancers with a high response rate to immunotherapy, such as melanoma and squamous cell lung cancer, have TMBs of 14.77 and 5.82 mutations/megabase, respectively (Appendix Figure A1A). Another marker, MSI-high, has been reported as a potential predictor of the effectiveness of immunotherapy in PDAC patients [31]. The TCGA mononucleotide and dinucleotide marker panel analysis found 24 PDAC patients with an indeterminate status, 115 with an MSS (microsatellite stable) status, 8 with an MSI-L (low MSI) status, and no patients with an MSI-H (high MSI) status (Appendix Figure A1B). Thus, it is difficult to apply TMB and MSI-H to measure immunotherapy responsiveness in PDAC.
The TME of PDAC is changeable, and modulation of the cell state can strongly influence drug responses, with state-specific vulnerabilities demonstrated in vivo and ex vivo [32]. Thus, we identified the proportions of 22 tumor-infiltrating lymphocytes in the TME of the TCGA PDAC patients using the CIBERSORT deconvolution algorithm. Consistent with previous studies, pro-tumor immune cells such as M0 and M2 macrophages and CD4+ memory resting cells were relatively abundant, but effector T cells such as CD4+ T cells and CD8+ T cells were relatively rare (Figure 5A), indicating a strong pro-tumor property and an immunosuppressive TME in PDAC. The results showed that most up-regulated UDEGs were significantly negatively correlated with the proportion of TME components associated with the response to ICB, including CD8+ T cells, naive B cells and activated dendritic cells, and significantly positively correlated with the proportion of TME components associated with resistance to ICB, including M0 macrophages, resting dendritic cells and Tregs (Figure 5B). Moreover, the down-UDEGs CYB5A and EPHX1 were significantly positively correlated with the proportion of CD8+ T cells, which have been reported to be associated with ICB responsiveness (Figure 5B). These results suggest that high expression of the up-regulated UDEGs and low expression of the down-regulated UDEGs are related to the formation of an immunosuppressive TME and might predict an unfavorable response to immunotherapy.

3. Discussion

The high heterogeneity between patients and the ubiquitous drug resistance are the main reasons for the poor efficacy of the current therapies for pancreatic cancer. Understanding the common molecular characteristics is essential to understanding the mechanisms of this universal therapeutic resistance and might be the key to overcoming this resistance. In this work, sixteen up-regulated and three down-regulated UDEGs were identified using paired cancer and normal samples and unpaired cancer bulk tissues. The single-cell analysis validated the high expression levels of the up-UDEGs and low levels of the down-UDEGs in cancer-related ductal cells compared to those in the control samples, which could represent the malignant changes in pancreatic cancer.
Among the nineteen UDEGs, other than the seven UDEGs reported to be carcinogenic and prometastatic genes involved in pancreatic cancer, the other UDEGs, such as ESM1, PCDH7, CORO2A, CEACAM5, GALNT5 and TMPRSS4, are also involved in other cancers. ESM1 (endothelial cell-specific molecule 1) can promote cancer progression and metastasis through the regulation of tumor cell proliferation, migration, invasion and drug resistance. PCDH7 (protocadherin 7) was reported to be related to cell growth, development and progression in prostate cancer [33], cervical cancer [34] and breast cancer [35]. CORO2A (coronin 2A) plays a critical role in cell migration and proliferation in breast cancer [36]. CEACAM5 (carcinoembryonic antigen-related cell adhesion molecule 5, also known as CEA) was reported to be a driver gene in colorectal cancer [37,38] and is related to tumor differentiation, invasion and metastasis [39]. GALNT5 (GalNAc transferase 5) mediates the carcinogenesis and progression of cholangiocarcinoma [40] and gastric cancer [41]. LRP8 is over-expressed in multiple cancers and is a potential therapeutic target [42,43,44,45]. TMPRSS4 is an important mediator of cell migration, invasion, epithelial–mesenchymal transition and metastasis in colon cancer cells, and increased TMPRSS4 expression correlated with colorectal cancer stage progression [46].
Based on these UDEGs, we performed a drug resistance and immune infiltration analysis and investigated the correlation between the expression of the UDEGs and the response to treatments for PDAC. These UDEGs were found to be closely related to the ubiquitous resistance of pancreatic cancer. For example, the higher expression of ANLN and GPRC5A was correlated with higher resistance to gemcitabine in pancreatic cell lines (ANLN: R = 0.43, p = 1.5 × 10−2; GPRCA5: R = 0.53, p = 2.3 × 10−3; Spearman correlation, Appendix Figure A2A). In contrast, in the TCGA PDAC cohorts, the non-responsive patients tended to express higher levels of ANLN and GPRC5A than the responsive patients (Appendix Figure A2B). Thus, PDAC patients with higher levels of ANLN and GPRC5A expression are more likely to be resistant to gemcitabine at the cell and tissue levels. Moreover, previous studies have shown that a down-regulation of ANLN can enhance the sensitivity of pancreatic cancer cells to gemcitabine [47] and inhibit doxorubicin resistance in human breast cancer cells [48]. Another study proved that knocking out GPRC5A suppressed the resistance to gemcitabine in a pancreatic cancer cell line (MIA PaCa-2) [19], and its over-expression enhanced the resistance of ovarian cancer cells [49]. SERPINB5 over-expression has been shown to promote metastasis in pancreatic ductal adenocarcinoma, and knockdown of SERPINB5 reduced the primary tumor weight and significantly decreased the amount of metastasis [24]. However, it has not been validated to be related to drug resistance and needs further validation through experiments. These results indicate that the UDEGs could be potential drug targets for pancreatic cancer. Thus, we also performed a DGI network analysis and found that some drugs used in other diseases interact with the UDEGs or their PPI partners that play important roles in the treatment of pancreatic cancer. These drugs are approved by the FDA and have passed preclinical testing, which not only ensures their safety but also greatly shortens the time for drug development and reduces the risk of failure.
In conclusion, we identified 19 universally differentially expressed genes (16 up-regulated and 3 down-regulated) in PDAC through a combination of bulk and single-cell analyses. These UDEGs are closely related to the drug resistance of PDAC, providing several potential targets for repurposing anticancer drugs to treat PDAC.

4. Materials and Methods

4.1. The Sources and Preprocessing of Expression Profiles

All the data used in our study were downloaded from public databases. Transcription profiles of PDAC and adjacent normal samples from PDAC patients and normal samples from pancreatic disease-free people came from the GEO (ncbi.nlm.nih.gov/geo, accessed on 13 March 2022), TCGA (cancergenome.nih.gov, accessed on 23 March 2022), GTEx (The Genotype-Tissue Expression, gtexportal.org, accessed on 13 March 2022) and ArrayExpress (ebi.ac.uk/arrayexpress, accessed on 13 March 2022) databases (Table 2).
For the data obtained using the Affymetrix platform, we applied the robust multi-array average algorithm for background adjustment without inner-sample normalization. Every probe was mapped to an Entrez gene ID according to the corresponding platform annotation file. The probes that mapped to multiple genes were discarded. If multiple probes were mapped to the same gene, the expression value of this gene was defined as the arithmetic mean of the values of the mapped probes. For the data sets obtained using the Illumina and Agilent platforms, we directly downloaded the processed expression data. For the profiles from the TCGA database generated using RNA-seq, the level 3 data were directly downloaded. Then, the Ensemble gene IDs were mapped to the Entrez gene IDs of protein-coding genes and used for further analysis. In this work, the PDAC and adjacent normal samples from the same patient were called paired samples; otherwise, they were referred to as unpaired samples.

4.2. Identification of Universally Dysregulated Genes in PDAC

We screened UDEGs using both paired and unpaired PDAC samples from public databases. In the paired samples, if the expression of a gene in the tumor sample was higher than that in the paired adjacent normal sample, it was defined as up-regulated and vice versa. For the unpaired samples, we screened dysregulated genes in individual samples by applying the RankComp method [11]. First, the normal background was derived from stable gene pairs, namely those gene pairs in which the rank of one gene is higher than or lower (Gi < Gj or Gi > Gj) than that of the other gene in more than 90% of the normal samples from different sources. Next, reverse gene pairs were defined for each disease sample as gene pairs with reversed rank ordering in comparison with their ordering in normal samples (Gi > Gj → Gi < Gj or Gi < Gj → Gi > Gj). Afterwards, Fisher’s exact test was used to test the null hypothesis that the numbers of reverse gene pairs supporting the up-regulation and down-regulation of a given gene (Gi) in a given disease sample (k) are equal. Based on the ratios of gene pairs with different ordering patterns in the normal and disease samples, we determined whether Gi is up-regulated (if the ratio in the disease sample is greater than in the normal samples) or down-regulated (if the ratio is smaller) or has stable expression (if the ratios are equal). A gene was determined to be a differentially expressed gene only if it remained significant after excluding the down-regulated (or up-regulated) partner genes from the reverse gene pairs supporting its up-regulation (or down-regulation) designation. Finally, the dysregulation frequency of each gene in the two approaches was calculated, and the overlapping genes with more than 85% frequency in both DEG lists were retained as UDEGs for the subsequent analyses.

4.3. Protein–Protein Interaction Network

A protein–protein interaction (PPI) network was created using PPIs from the STRING database [50]. Each PPI is annotated with a confidence score, which represents how likely STRING judges an interaction to be true given the available evidence. The scores range from 0 to 1, with 1 representing the highest possible confidence. To ensure the high confidence of the PPIs used to generate our network, we set the interaction score threshold to 0.99.

4.4. Prognostic Efficiency and Functions of UDEGs

A survival analysis was performed on the TCGA PDAC cohort using the R package “survival” (version: R 4.0.2)to explore the prognostic efficacy of UDEGs. The CancerMine [51] database was searched to explore the reported roles of the UDEGs in multiple cancers. To analyze the functions of the UDEGs, a KEGG pathway analysis was conducted, and REACTOME [52] (reactome.org, accessed on 20 May 2022) was used to systematically display the hierarchy of the pathway enrichment in this study.

4.5. Processing of scRNA-Seq Data

The raw scRNA-seq data were downloaded from the GSA database (Genome Sequence Archive, ngdc.cncb.ac.cn/gsa, accessed on 21 April 2022, CRA001160 [53]). The “Seurat” [54] (v2.3.0) R package was used for quality control and downstream single-cell analysis. For quality control, we set the following criteria: <200 genes/cell, <3 cells/gene and >10% mitochondrial genes/cell. The tumor and control samples were merged and scaled.

4.6. Cluster Identification and Cell Type Assignment

The top 2000 variable genes were used for clustering. T-SNE reduction was applied to display the clustering results. The cluster-specific marker genes were identified by running the “FindAllMarkers” function of the “Seurat” package. We used some well-known markers from the CellMarker database [55] and the literature to identify specific cell types; for example, we used CPA1, PRSS1, CTRB1, CTRB2 and REG1B to identify acinar cells; MS4A1 and CD27 to identify B cells; AIF1 and CD68 to identify macrophages; PCSK1N, CHGB, CHGA, INS and SLC30A8 to identify endocrine cells; KRT19, MMP7, TSPAN8, SOX9 and LCN2 to identify ductal cells; MUC1 and PROM1 to identify cancer cells; LUM, DCN and LRP1 to identify fibroblasts; MNDA and ITGB2 to identify monocytes; RGS5, ACTA2, PDGFRB and ADIRF to identify stellate cells; and CD2, CD3D and CD3E to identify T cells. Using Seurat’s FindAllMarkers function, we identified genes that were highly expressed in acinar cells, duct cells, macrophages, endocrine cells, fibroblasts, endothelial cells, monocytes, stellate cells and T cells in pancreatic cancer and normal tissues. Then, KEGG functional enrichment analysis was performed on each gene list (p <= 0.05). The functional conformance of common cell types between the pancreatic and normal tissue samples was determined using the following formula:
C o n s i s t e n c e = ( c o n n + c o n t ) / 2
For a given cell type, n is the number of pathways enriched with the DEGs of this cell in the normal tissues, t is the number of pathways enriched with the DEGs of this cell in the cancer tissues and con is the number of common pathways in both the cancer and normal tissues.

4.7. Drug Resistance

The processed expression profiles and sensitivity data of 32 pancreatic cell lines were downloaded from the GDSC database (Genomics of Drug Sensitivity in Cancer [56], cancerrxgene.org, accessed on 21 June 2022). The sensitivity of a cell line to a drug is represented by the IC50 value. For each drug–gene pair, we calculated the Spearman correlation coefficient between the expression values of the gene and the IC50 values of the drug across different cell lines. Only the drugs that were effective against at least half of the 32 cell lines and had drug–gene correlations with a p-value < 0.05 were retained.

4.8. Drug–Gene Interaction Network

We explored the potential of the UDEGs to be drug targets by constructing a drug–gene interaction (DGI) network using information from DGIdb [57] (dgidb.genome.wustl.edu, accessed on 23 June 2022). We select drug–gene interactions with scores greater than 5 as candidate interactions and obtained the drugs’ FDA approval information, mechanism and function from the DrugBank database (go.drugbank.com/drugs, accessed on 23 June 2022).

4.9. TMB and MSI Information

The “TCGAmutations” [58] R package was used to calculate the tumor mutation burden (TMB) of the TCGA PDAC, SKCM (skin cutaneous melanoma) and LUSC (lung squamous cell carcinoma) patients. The “TCGAbiolinks” R package was used to download the microsatellite instability (MSI) information of the TCGA patients.

4.10. Immune Cell Proportion of TCGA PDAC Patients

CIBERSORT [59] was applied to obtain the proportions of 22 immune cell types, including macrophages (M0, M1 and M2), CD8+ T cells, naive CD4+ T cells, CD4+ memory resting T cells, CD4+ memory activated T cells, monocytes, resting dendritic cells, neutrophils, resting mast cells, naive B cells, eosinophils, memory B cells, resting NK cells, activated NK cells, follicular helper T cells, regulatory T cells (Tregs), gamma delta T cells, activated dendritic cells, plasma cells and activated mast cells. The correlations between the expression of the UDEGs and cell proportions were calculated using Spearman correlation.

4.11. Statistical Analysis

All statistical analyses were performed using R 4.0.2.

Author Contributions

H.Y.: writing—review and editing, writing—original draft, supervision, methodology, funding acquisition, conceptualization. J.X.: writing—review and editing, writing—original draft, visualization, validation, methodology, funding acquisition, data curation, conceptualization. L.Z.: writing—review and editing, methodology, data curation. H.Z.: writing—review and editing, software, resources, methodology, investigation. Q.F.: writing—review and editing, methodology. H.L.: writing—review and editing, methodology. O.W.: writing—review and editing, investigation, data curation. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Natural Science Foundation of Fujian Province [grant number 2023J01755] and the Scientific Research Startup Fund of Guizhou Medical University [grant number 26232020141].

Institutional Review Board Statement

This study solely relies on data retrieved from public databases. Upon careful evaluation, the data we utilized have already met all the necessary ethical and legal standards during their initial collection and curation in the respective public databases. Our research strictly involves secondary analysis of publicly available, de-identified data. Thus, no new ethical issues are introduced by our data - usage approach. Consequently, this research is exempt from the need for ethical review, whether it pertains to animal or human ethics.

Data Availability Statement

The original data presented in the study are openly available in Gene Expression Omnibus (GEO, ncbi.nlm.nih.gov/geo), The Cancer Genome Atlas (TCGA, cancergenome.nih.gov), The Genotype-Tissue Expression (GTEX, gtexportal.org), ArrayExpress (ebi.ac.uk/arrayexpress), GSA database (Genome Sequence Archive, ngdc.cncb.ac.cn/gsa), Genomics of Drug Sensitivity in Cancer (GDSC, cancerrxgene.org), DGIdb (dgidb.genome.wustl.edu), and DrugBank database (go.drugbank.com/drugs).

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

UDEGsuniversally differentially expressed genes
PDACpancreatic ductal adenocarcinoma
ICB immune checkpoint blockade
TME tumor microenvironment
DEG differentially expressed gene
PPI protein–protein interaction
DGIdrug–gene interaction
TMB tumor mutation burden
MSI microsatellite instability
GEOGene Expression Omnibus
TCGAThe Cancer Genome Atlas
GTEXThe Genotype-Tissue Expression
GDSCGenomics of Drug Sensitivity in Cancer

Appendix A

Figure A1. MSI counts and TMB in PDAC. (A) MSI statistics for TCGA PDAC patients. (B) Comparison of TMB between TCGA PDAC, LUSC and SKCM patients. MSI-H: high microsatellite instability; MSI-L: low microsatellite instability; MSS: microsatellite stability; PDAC: pancreatic ductal adenocarcinoma; SKCM: skin cutaneous melanoma; LUSC: lung squamous cell carcinoma.
Figure A1. MSI counts and TMB in PDAC. (A) MSI statistics for TCGA PDAC patients. (B) Comparison of TMB between TCGA PDAC, LUSC and SKCM patients. MSI-H: high microsatellite instability; MSI-L: low microsatellite instability; MSS: microsatellite stability; PDAC: pancreatic ductal adenocarcinoma; SKCM: skin cutaneous melanoma; LUSC: lung squamous cell carcinoma.
Ijms 26 03936 g0a1
Figure A2. Relationship between resistance to gemcitabine and expression of ANLN and GPRC5A. (A) Expression of ANLN and GPRC5A in TCGA PDAC patient groups with different responses to gemcitabine. (B) Number of significant correlations between drug resistance and UDEG expression.
Figure A2. Relationship between resistance to gemcitabine and expression of ANLN and GPRC5A. (A) Expression of ANLN and GPRC5A in TCGA PDAC patient groups with different responses to gemcitabine. (B) Number of significant correlations between drug resistance and UDEG expression.
Ijms 26 03936 g0a2
Table A1. Functions of UDEGs in cancers.
Table A1. Functions of UDEGs in cancers.
GeneCancer TypeRole
CEACAM5colorectal cancerDriver
ESM1esophageal cancerOncogene
SFNprostate cancer, kidney cancer, pancreatic cancer, lung adenocarcinoma, malignant glioma, osteosarcomaOncogene
SFNbreast cancer, tongue squamous cell carcinomaTumor_Suppressor
LAMC2pancreatic cancer, penis squamous cell carcinoma, esophagus squamous cell carcinoma, ovarian cancerOncogene
PCDH7colon cancerOncogene
SERPINB5breast cancer, stomach cancer, breast carcinomaOncogene
SERPINB5breast cancer, prostate cancer, squamous cell carcinoma, melanoma, breast carcinoma, colorectal cancer, urinary bladder cancer, transitional cell carcinoma, stomach cancer, laryngeal carcinoma, lung squamous cell carcinoma, invasive lobular carcinoma, lung adenocarcinomaTumor_Suppressor
ANLNpancreatic cancerOncogene
CORO2Abreast cancerOncogene
GPRC5Alung non-small cell carcinoma, lung cancer, head and neck squamous cell carcinoma, stomach cancer, breast cancerTumor_Suppressor
GPRC5Apancreatic ductal adenocarcinoma, pancreatic cancer, prostate cancerOncogene
ACADMbreast cancerOncogene
Table A2. 371 significantly correlated drug–gene relations.
Table A2. 371 significantly correlated drug–gene relations.
DrugGeneThresholdDrugGeneThreshold
CAMPTOTHECINGPRC5APositiveOSU-03012SERPINB5Positive
VINBLASTINEGPRC5APositiveEMBELINANLNPositive
VINBLASTINESERPINB5PositiveEMBELINGPRC5APositive
CISPLATINANLNPositiveFH535GPRC5APositive
CISPLATINGPRC5APositiveFH535LRP8Negative
CISPLATINSERPINB5PositiveFH535SDR16C5Positive
CYTARABINEANLNPositiveFH535SERPINB5Positive
CYTARABINEGPRC5APositiveIPA-3ANLNPositive
CYTARABINESERPINB5PositiveIPA-3SERPINB5Positive
GEFITINIBGPRC5APositiveGSK650394ANLNPositive
NILOTINIBGPRC5APositiveGSK650394SERPINB5Positive
OLAPARIBANLNPositiveBAY-61-3606SERPINB5Positive
OLAPARIBGPRC5APositiveTHAPSIGARGINCORO2APositive
AZD7762LRP8NegativeBEXAROTENEGPRC5APositive
AZD7762SERPINB5NegativeBEXAROTENESERPINB5Positive
AZD7762SFNNegativeBEXAROTENESFNPositive
AFATINIBACADMPositiveBLEOMYCINANLNPositive
STAUROSPORINEGPRC5APositiveBLEOMYCINSERPINB5Positive
NUTLIN-3A (-)GPRC5APositiveLFM-A13GPRC5APositive
PD173074ANLNPositiveGW-2580ANLNPositive
PD173074GPRC5APositivePHENFORMINANLNPositive
PD173074LRP8NegativePHENFORMINSERPINB5Positive
PALBOCICLIBGPRC5APositivePAZOPANIBCORO2APositive
PALBOCICLIBSERPINB5PositivePAZOPANIBLRP8Negative
PD0325901PCDH7PositiveDACINOSTATANLNPositive
DASATINIBPCDH7PositiveEPOTHILONE BANLNPositive
PACLITAXELACADMPositiveEPOTHILONE BSERPINB5Positive
PACLITAXELGPRC5APositiveTIPIFARNIBANLNPositive
PACLITAXELPCDH7PositiveAVAGACESTATANLNPositive
RAPAMYCINGPRC5APositiveAS601245SERPINB5Positive
RAPAMYCINLRP8NegativeISPINESIB MESYLATESERPINB5Positive
RAPAMYCINPCDH7PositiveTL-2-105ANLNPositive
SORAFENIBANLNPositiveTL-2-105SERPINB5Positive
SORAFENIBGPRC5APositiveTAK-715ANLNPositive
SORAFENIBSERPINB5PositiveBX-912ANLNPositive
IRINOTECANGPRC5APositiveBX-912SERPINB5Positive
GSK1904529AGPRC5APositiveAS605240SERPINB5Positive
ERLOTINIBCORO2ANegativeGENENTECH CPD 10SERPINB5Positive
ERLOTINIBSDR16C5NegativeGSK1070916ANLNPositive
MK-1775GPRC5APositiveGSK1070916SERPINB5Positive
MK-1775LRP8NegativeENZASTAURINPCDH7Negative
DINACICLIBLRP8NegativeGSK429286AANLNPositive
GEMCITABINEANLNPositiveGSK429286AGPRC5APositive
GEMCITABINEGPRC5APositiveGSK429286ASERPINB5Positive
TAMOXIFENLRP8NegativeGSK429286ASFNPositive
EPZ004777LRP8NegativeQL-XII-47SERPINB5Positive
YK-4-279GPRC5APositiveIC-87114SERPINB5Positive
YK-4-279SERPINB5PositiveUNC0638SERPINB5Positive
DAPORINADCORO2APositiveXMD14-99ANLNPositive
DAPORINADSERPINB5PositiveXMD14-99SERPINB5Positive
TALAZOPARIBANLNPositiveCP724714ANLNPositive
TALAZOPARIBGPRC5APositiveCP724714SERPINB5Positive
XAV939ACADMPositiveJW-7-24-1SERPINB5Positive
DABRAFENIBANLNPositiveNPK76-II-72-1ANLNPositive
DABRAFENIBGPRC5APositiveNG-25ANLNPositive
DABRAFENIBLRP8NegativeTL-1-85ANLNPositive
TEMOZOLOMIDEGPRC5APositiveACY-1215ANLNPositive
TEMOZOLOMIDELRP8NegativeACY-1215LRP8Positive
IAP_5620GPRC5APositiveACY-1215SERPINB5Positive
AZD1208GPRC5APositiveACY-1215SFNPositive
EPIRUBICINANLNPositiveTUBASTATIN AANLNPositive
EPIRUBICINGPRC5APositiveTUBASTATIN AGPRC5APositive
CYCLOPHOSPHAMIDEGPRC5APositiveTUBASTATIN ASERPINB5Positive
PEVONEDISTATANLNPositiveZIBOTENTANANLNPositive
PEVONEDISTATSERPINB5PositiveZIBOTENTANGPRC5APositive
SAPITINIBSDR16C5NegativeZIBOTENTANSERPINB5Positive
SAPITINIBSERPINB5NegativeNSC-207895ANLNPositive
SAPITINIBSFNNegativeVNLG/124SERPINB5Positive
LCL161SERPINB5NegativeAR-42ANLNPositive
LAPATINIBGPRC5APositiveAR-42SERPINB5Positive
LAPATINIBLRP8NegativeCUDC-101ANLNPositive
LUMINESPIBCORO2APositiveCUDC-101SERPINB5Positive
ALPELISIBACADMPositiveBELINOSTATANLNPositive
TASELISIBACADMPositiveBELINOSTATSERPINB5Positive
EPZ5676GPRC5APositiveCAY10603ANLNPositive
IWP-2LRP8NegativeCAY10603SERPINB5Positive
LGK974CORO2ANegativeJNJ38877605LRP8Positive
WZ4003LRP8NegativeSU11274SERPINB5Positive
CZC24832GPRC5APositiveKIN001-236ANLNPositive
CZC24832LRP8NegativeKIN001-260ANLNPositive
AZD5582LRP8NegativeNVP-BHG712ANLNPositive
GSK2606414CORO2ANegativeOSI-930ANLNPositive
PFI3PCDH7PositiveOSI-930SERPINB5Positive
PCI-34051LRP8NegativeCX-5461ANLNPositive
PCI-34051PCDH7PositiveCX-5461SERPINB5Positive
WNT-C59SERPINB5NegativePHA-793887SERPINB5Positive
I-BET-762ANLNPositivePI-103CORO2APositive
RVX-208PCDH7PositiveFEDRATINIBANLNPositive
GSK343ACADMPositiveY-39983ANLNPositive
ML323ANLNPositiveY-39983GPRC5APositive
ML323GPRC5APositiveY-39983SERPINB5Positive
PRT062607LRP8NegativeYM201636ANLNPositive
AGI-6780SFNNegativeWYE-125132PCDH7Negative
CDK9_5576LRP8NegativeGSK690693SERPINB5Positive
CDK9_5038LRP8NegativeSNX-2112ANLNPositive
EG5_9814GPRC5APositiveSNX-2112GPRC5APositive
EG5_9814LRP8NegativeXMD13-2ANLNPositive
EG5_9814SERPINB5PositiveXMD13-2SERPINB5Positive
ERK_2440LRP8NegativeQL-X-138SERPINB5Positive
ERK_2440SDR16C5NegativeXMD15-27ANLNPositive
AZD5991GPRC5APositiveT0901317GPRC5APositive
AZD5991LRP8NegativeSELISISTATSERPINB5Positive
PAK_5339LRP8NegativeKIN001-270SERPINB5Positive
TAF1_5496ACADMNegativeTHZ-2-102-1ANLNPositive
TAF1_5496ANLNPositiveAT7867SERPINB5Positive
TAF1_5496GPRC5APositiveAT7867SFNPositive
SELUMETINIBGPRC5ANegativeCI-1033PCDH7Negative
TENIPOSIDEANLNPositiveTWS119GPRC5ANegative
TENIPOSIDEGPRC5APositiveTWS119LRP8Positive
TENIPOSIDELRP8NegativeTORIN 2PCDH7Negative
MITOXANTRONEGPRC5APositivePILARALISIBSERPINB5Positive
MITOXANTRONELRP8NegativeGSK1059615ANLNPositive
DACTINOMYCINPCDH7PositiveBRIVANIB, BMS-540215SERPINB5Positive
NELARABINEACADMPositiveBIBF-1120CORO2ANegative
NELARABINEGPRC5APositiveAST-1306CORO2ANegative
VINCRISTINEGPRC5APositiveAST-1306SDR16C5Negative
VINCRISTINELRP8NegativeAPITOLISIBLRP8Positive
PODOPHYLLOTOXIN BROMIDEGPRC5APositiveAPITOLISIBPCDH7Negative
PODOPHYLLOTOXIN BROMIDELRP8NegativeLIMK1 INHIBITOR BMS4ACADMNegative
DIHYDROROTENONEANLNPositiveKB NB 142-70CORO2APositive
ELEPHANTINLRP8NegativeSPHINGOSINE KINASE 1 INHIBITOR IISERPINB5Positive
SABUTOCLAXCORO2APositiveEEF2K INHIBITOR, A-484954SERPINB5Positive
MG-132GPRC5APositiveARA-GGPRC5APositive
MG-132PCDH7PositiveC-75SERPINB5Positive
BDP-00009066GPRC5APositiveCAP-232, TT-232, TLN-232GPRC5APositive
BDP-00009066LRP8NegativeTRICHOSTATIN AANLNPositive
BUPARLISIBGPRC5APositiveTRICHOSTATIN AGPRC5APositive
ULIXERTINIBANLNPositivePANOBINOSTATANLNPositive
ULIXERTINIBLRP8NegativeIMD-0354SERPINB5Positive
AFURESERTIBGPRC5APositiveETP-45835SERPINB5Positive
AFURESERTIBLRP8NegativeCD532ANLNPositive
AFURESERTIBPCDH7PositiveCD532SERPINB5Positive
AGI-5198ANLNPositiveARRY-520SERPINB5Positive
AGI-5198GPRC5APositiveSB505124GPRC5APositive
AZD3759SDR16C5NegativeA-83-01LRP8Positive
AZD5363GPRC5APositiveFTY-720GPRC5APositive
AZD5363LRP8NegativeKOBE2602SERPINB5Positive
IPATASERTIBLRP8NegativeTRETINOINGPRC5APositive
IPATASERTIBPCDH7PositiveCI-1040SDR16C5Positive
GDC0810GPRC5APositiveTEMSIROLIMUSACADMNegative
GSK2578215AGPRC5APositiveTEMSIROLIMUSSDR16C5Positive
GSK2578215ALRP8NegativeTEMSIROLIMUSSERPINB5Positive
GSK2578215APCDH7PositiveBOSUTINIBACADMNegative
I-BRD9LRP8NegativeAXITINIBGPRC5APositive
I-BRD9PCDH7PositiveAXITINIBSERPINB5Positive
TELOMERASE INHIBITOR IXPCDH7PositiveGW441756SERPINB5Positive
MIRA-1GPRC5APositiveVX-702ANLNPositive
MIRA-1LRP8NegativeELESCLOMOLACADMNegative
SAVOLITINIBANLNPositiveELESCLOMOLSDR16C5Positive
UMI-77PCDH7PositiveVISMODEGIBANLNPositive
SEPANTRONIUM BROMIDEANLNPositiveVISMODEGIBSERPINB5Positive
SEPANTRONIUM BROMIDECORO2APositiveBX795SDR16C5Positive
MIM1GPRC5APositiveBX795SERPINB5Positive
BPD-00008900GPRC5APositiveSL0101ANLNPositive
BPD-00008900PCDH7PositiveSL0101SERPINB5Positive
BPD-00008900SERPINB5PositiveJNK INHIBITOR VIIILRP8Negative
FORETINIBANLNPositiveSB590885ANLNPositive
BIBR-1532ANLNPositiveCETUXIMABGPRC5ANegative
PYRIDOSTATINGPRC5APositiveHG-5-88-01LRP8Positive
PYRIDOSTATINPCDH7PositiveTW 37SERPINB5Positive
AMG-319GPRC5APositiveTW 37SFNPositive
AMG-319LRP8NegativeXMD8-92LRP8Positive
MK-8776GPRC5APositiveQL-VIII-58PCDH7Negative
VINORELBINEANLNPositiveCCT-018159CORO2APositive
VINORELBINEGPRC5APositiveCCT-018159SERPINB5Positive
VINORELBINESERPINB5PositiveCCT-018159SFNPositive
VX-11EANLNPositiveQL-XII-61GPRC5ANegative
VX-11ELRP8NegativeIOX2SERPINB5Positive
LJI308LRP8Negative(5Z)-7-OXOZEAENOLSDR16C5Positive
AT13148GPRC5APositiveUNC1215SERPINB5Positive
AT13148LRP8NegativeUNC0642CORO2APositive
GSK269962AANLNPositiveUNC0642SERPINB5Positive
GSK269962AGPRC5APositiveSGC0946SERPINB5Positive
GSK269962ASERPINB5PositiveICL1100013CORO2APositive
DOXORUBICINANLNPositiveICL1100013SERPINB5Positive
DOXORUBICINSERPINB5PositiveAZD4877SERPINB5Positive
ETOPOSIDEANLNPositiveEPHB4_9721ACADMPositive
ETOPOSIDESERPINB5PositiveEPHB4_9721GPRC5ANegative
MITOMYCIN-CANLNPositiveEPHB4_9721SFNNegative
MITOMYCIN-CSERPINB5PositiveBPTESLRP8Positive
CP466722SERPINB5PositiveIGFR_3801LRP8Positive
PONATINIBLRP8NegativeJAK1_3715SERPINB5Positive
PF-562271SERPINB5PositivePARP_0108GPRC5APositive
DMOGGPRC5ANegativePARP_9482GPRC5APositive
OSU-03012ANLNPositiveTANK_1366GPRC5ANegative
OSU-03012GPRC5APositive

References

  1. Bliss, L.A.; Witkowski, E.R.; Yang, C.J.; Tseng, J.F. Outcomes in operative management of pancreatic cancer. J. Surg. Oncol. 2014, 110, 592–598. [Google Scholar] [CrossRef] [PubMed]
  2. Conroy, T.; Hammel, P.; Hebbar, M.; Ben Abdelghani, M.; Wei, A.C.; Raoul, J.L.; Chone, L.; Francois, E.; Artru, P.; Biagi, J.J.; et al. FOLFIRINOX or Gemcitabine as Adjuvant Therapy for Pancreatic Cancer. N. Engl. J. Med. 2018, 379, 2395–2406. [Google Scholar] [CrossRef] [PubMed]
  3. He, J.; Blair, A.B.; Groot, V.P.; Javed, A.A.; Burkhart, R.A.; Gemenetzis, G.; Hruban, R.H.; Waters, K.M.; Poling, J.; Zheng, L.; et al. Is a Pathological Complete Response Following Neoadjuvant Chemoradiation Associated With Prolonged Survival in Patients With Pancreatic Cancer? Ann. Surg. 2018, 268, 1–8. [Google Scholar] [CrossRef] [PubMed]
  4. Tang, K.; Lu, W.; Qin, W.; Wu, Y. Neoadjuvant therapy for patients with borderline resectable pancreatic cancer: A systematic review and meta-analysis of response and resection percentages. Pancreatol. Off. J. Int. Assoc. Pancreatol. 2016, 16, 28–37. [Google Scholar] [CrossRef]
  5. Chuong, M.D.; Springett, G.M.; Freilich, J.M.; Park, C.K.; Weber, J.M.; Mellon, E.A.; Hodul, P.J.; Malafa, M.P.; Meredith, K.L.; Hoffe, S.E.; et al. Stereotactic body radiation therapy for locally advanced and borderline resectable pancreatic cancer is effective and well tolerated. Int. J. Radiat. Oncol. Biol. Phys. 2013, 86, 516–522. [Google Scholar] [CrossRef]
  6. Moutardier, V.; Magnin, V.; Turrini, O.; Viret, F.; Hennekinne-Mucci, S.; Goncalves, A.; Pesenti, C.; Guiramand, J.; Lelong, B.; Giovannini, M.; et al. Assessment of pathologic response after preoperative chemoradiotherapy and surgery in pancreatic adenocarcinoma. Int. J. Radiat. Oncol. Biol. Phys. 2004, 60, 437–443. [Google Scholar] [CrossRef]
  7. Chuong, M.D.; Frakes, J.M.; Figura, N.; Hoffe, S.E.; Shridhar, R.; Mellon, E.A.; Hodul, P.J.; Malafa, M.P.; Springett, G.M.; Centeno, B.A. Histopathologic tumor response after induction chemotherapy and stereotactic body radiation therapy for borderline resectable pancreatic cancer. J. Gastrointest. Oncol. 2016, 7, 221–227. [Google Scholar] [CrossRef]
  8. Morrison, A.H.; Byrne, K.T.; Vonderheide, R.H. Immunotherapy and Prevention of Pancreatic Cancer. Trends Cancer 2018, 4, 418–428. [Google Scholar] [CrossRef]
  9. Royal, R.E.; Levy, C.; Turner, K.; Mathur, A.; Hughes, M.; Kammula, U.S.; Sherry, R.M.; Topalian, S.L.; Yang, J.C.; Lowy, I.; et al. Phase 2 trial of single agent Ipilimumab (anti-CTLA-4) for locally advanced or metastatic pancreatic adenocarcinoma. J. Immunother. 2010, 33, 828–833. [Google Scholar] [CrossRef]
  10. Kim, J.M.; Chen, D.S. Immune escape to PD-L1/PD-1 blockade: Seven steps to success (or failure). Ann. Oncol. 2016, 27, 1492–1504. [Google Scholar] [CrossRef]
  11. Wang, H.; Sun, Q.; Zhao, W.; Qi, L.; Gu, Y.; Li, P.; Zhang, M.; Li, Y.; Liu, S.L.; Guo, Z. Individual-level analysis of differential expression of genes and pathways for personalized medicine. Bioinformatics 2015, 31, 62–68. [Google Scholar] [CrossRef] [PubMed]
  12. Shema, E.; Bernstein, B.E.; Buenrostro, J.D. Single-cell and single-molecule epigenomics to uncover genome regulation at unprecedented resolution. Nat. Genet. 2019, 51, 19–25. [Google Scholar] [CrossRef] [PubMed]
  13. Lawson, D.A.; Kessenbrock, K.; Davis, R.T.; Pervolarakis, N.; Werb, Z. Tumour heterogeneity and metastasis at single-cell resolution. Nat. Cell Biol. 2018, 20, 1349–1360. [Google Scholar] [CrossRef]
  14. Sato, N.; Maitra, A.; Fukushima, N.; van Heek, N.T.; Matsubayashi, H.; Iacobuzio-Donahue, C.A.; Rosty, C.; Goggins, M. Frequent hypomethylation of multiple genes overexpressed in pancreatic ductal adenocarcinoma. Cancer Res. 2003, 63, 4158–4166. [Google Scholar]
  15. Fukushige, S.; Horii, A. Road to early detection of pancreatic cancer: Attempts to utilize epigenetic biomarkers. Cancer Lett. 2014, 342, 231–237. [Google Scholar] [CrossRef]
  16. Pyke, C.; Romer, J.; Kallunki, P.; Lund, L.R.; Ralfkiaer, E.; Dano, K.; Tryggvason, K. The gamma 2 chain of kalinin/laminin 5 is preferentially expressed in invading malignant cells in human cancers. Am. J. Pathol. 1994, 145, 782–791. [Google Scholar] [PubMed]
  17. Wang, H.; Cai, J.; Du, S.; Wei, W.; Shen, X. LAMC2 modulates the acidity of microenvironments to promote invasion and migration of pancreatic cancer cells via regulating AKT-dependent NHE1 activity. Exp. Cell Res. 2020, 391, 111984. [Google Scholar] [CrossRef]
  18. Kosanam, H.; Prassas, I.; Chrystoja, C.C.; Soleas, I.; Chan, A.; Dimitromanolakis, A.; Blasutig, I.M.; Ruckert, F.; Gruetzmann, R.; Pilarsky, C.; et al. Laminin, gamma 2 (LAMC2): A promising new putative pancreatic cancer biomarker identified by proteomic analysis of pancreatic adenocarcinoma tissues. Mol. Cell. Proteom. MCP 2013, 12, 2820–2832. [Google Scholar] [CrossRef]
  19. Liu, B.; Yang, H.; Pilarsky, C.; Weber, G.F. The Effect of GPRC5a on the Proliferation, Migration Ability, Chemotherapy Resistance, and Phosphorylation of GSK-3beta in Pancreatic Cancer. Int. J. Mol. Sci. 2018, 19, 1870. [Google Scholar] [CrossRef]
  20. Rozengurt, E.; Sinnett-Smith, J.; Eibl, G. Yes-associated protein (YAP) in pancreatic cancer: At the epicenter of a targetable signaling network associated with patient survival. Signal Transduct. Target. Ther. 2018, 3, 11. [Google Scholar] [CrossRef]
  21. Zhou, S.; Yan, Y.; Chen, X.; Zeng, S.; Wei, J.; Wang, X.; Gong, Z.; Xu, Z. A two-gene-based prognostic signature for pancreatic cancer. Aging 2020, 12, 18322–18342. [Google Scholar] [CrossRef] [PubMed]
  22. Wang, A.; Dai, H.; Gong, Y.; Zhang, C.; Shu, J.; Luo, Y.; Jiang, Y.; Liu, W.; Bie, P. ANLN-induced EZH2 upregulation promotes pancreatic cancer progression by mediating miR-218-5p/LASP1 signaling axis. J. Exp. Clin. Cancer Res. 2019, 38, 347. [Google Scholar] [CrossRef]
  23. Lin, L.Y.; Yeh, Y.C.; Chu, C.H.; Won, J.G.S.; Shyr, Y.M.; Chao, Y.; Li, C.P.; Wang, S.E.; Chen, M.H. Endocan expression is correlated with poor progression-free survival in patients with pancreatic neuroendocrine tumors. Medicine 2017, 96, e8262. [Google Scholar] [CrossRef]
  24. Tian, C.; Ohlund, D.; Rickelt, S.; Lidstrom, T.; Huang, Y.; Hao, L.; Zhao, R.T.; Franklin, O.; Bhatia, S.N.; Tuveson, D.A.; et al. Cancer Cell-Derived Matrisome Proteins Promote Metastasis in Pancreatic Ductal Adenocarcinoma. Cancer Res. 2020, 80, 1461–1474. [Google Scholar] [CrossRef]
  25. Penheiter, A.R.; Erdogan, S.; Murphy, S.J.; Hart, S.N.; Felipe Lima, J.; Rakhshan Rohakhtar, F.; O’Brien, D.R.; Bamlet, W.R.; Wuertz, R.E.; Smyrk, T.C.; et al. Transcriptomic and Immunohistochemical Profiling of SLC6A14 in Pancreatic Ductal Adenocarcinoma. Biomed. Res. Int. 2015, 2015, 593572. [Google Scholar] [CrossRef]
  26. Coothankandaswamy, V.; Cao, S.; Xu, Y.; Prasad, P.D.; Singh, P.K.; Reynolds, C.P.; Yang, S.; Ogura, J.; Ganapathy, V.; Bhutia, Y.D. Amino acid transporter SLC6A14 is a novel and effective drug target for pancreatic cancer. Br. J. Pharmacol. 2016, 173, 3292–3306. [Google Scholar] [CrossRef] [PubMed]
  27. Li, Y.; Zhu, Y.Y.; Dai, G.P.; Wu, D.J.; Gao, Z.Z.; Zhang, L.; Fan, Y.H. Screening and validating the core biomarkers in patients with pancreatic ductal adenocarcinoma. Math. Biosci. Eng. 2019, 17, 910–927. [Google Scholar] [CrossRef]
  28. Cortes, E.; Sarper, M.; Robinson, B.; Lachowski, D.; Chronopoulos, A.; Thorpe, S.D.; Lee, D.A.; Del Rio Hernandez, A.E. GPER is a mechanoregulator of pancreatic stellate cells and the tumor microenvironment. EMBO Rep. 2019, 20, e46556. [Google Scholar] [CrossRef] [PubMed]
  29. Li, B.; Yang, J.; Lu, Z.; Liu, B.; Liu, F. A study on the mechanism of rapamycin mediating the sensitivity of pancreatic cancer cells to cisplatin through PI3K/AKT/mTOR signaling pathway. J. BUON 2019, 24, 739–745. [Google Scholar]
  30. Spencer, A.; Yoon, S.S.; Harrison, S.J.; Morris, S.R.; Smith, D.A.; Brigandi, R.A.; Gauvin, J.; Kumar, R.; Opalinska, J.B.; Chen, C. The novel AKT inhibitor afuresertib shows favorable safety, pharmacokinetics, and clinical activity in multiple myeloma. Blood 2014, 124, 2190–2195. [Google Scholar] [CrossRef]
  31. Le, D.T.; Durham, J.N.; Smith, K.N.; Wang, H.; Bartlett, B.R.; Aulakh, L.K.; Lu, S.; Kemberling, H.; Wilt, C.; Luber, B.S.; et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science 2017, 357, 409–413. [Google Scholar] [CrossRef] [PubMed]
  32. Raghavan, S.; Winter, P.S.; Navia, A.W.; Williams, H.L.; DenAdel, A.; Lowder, K.E.; Galvez-Reyes, J.; Kalekar, R.L.; Mulugeta, N.; Kapner, K.S.; et al. Microenvironment drives cell state, plasticity, and drug response in pancreatic cancer. Cell 2021, 184, 6119–6137.e26. [Google Scholar] [CrossRef]
  33. Terry, S.; Queires, L.; Gil-Diez-de-Medina, S.; Chen, M.W.; de la Taille, A.; Allory, Y.; Tran, P.L.; Abbou, C.C.; Buttyan, R.; Vacherot, F. Protocadherin-PC promotes androgen-independent prostate cancer cell growth. Prostate 2006, 66, 1100–1113. [Google Scholar] [CrossRef] [PubMed]
  34. Zhang, S.; Fu, X. The Clinical Significance and Biological Function of PCDH7 in Cervical Cancer. Cancer Manag. Res. 2021, 13, 3841–3847. [Google Scholar] [CrossRef] [PubMed]
  35. Cao, J.; Wang, M.; Wang, T. CCAAT enhancer binding protein beta has a crucial role in regulating breast cancer cell growth via activating the TGF-beta-Smad3 signaling pathway. Exp. Ther. Med. 2017, 14, 1554–1560. [Google Scholar] [CrossRef]
  36. Deng, J.L.; Zhang, H.B.; Zeng, Y.; Xu, Y.H.; Huang, Y.; Wang, G. Effects of CORO2A on Cell Migration and Proliferation and Its Potential Regulatory Network in Breast Cancer. Front. Oncol. 2020, 10, 916. [Google Scholar] [CrossRef]
  37. Ye, L.; Yang, Y.; Ma, X.Y.; Li, D.; Xu, M.L.; Tan, P.; Long, L.M.; Wang, H.Q.; Liu, T.; Guo, Y.H. Construction of a novel vector expressing Survivin-shRNA and fusion suicide gene yCDglyTK and its application in inhibiting proliferation and migration of colon cancer cells. Exp. Ther. Med. 2017, 14, 4721–4728. [Google Scholar] [CrossRef] [PubMed]
  38. Lee, J.K.; Bangayan, N.J.; Chai, T.; Smith, B.A.; Pariva, T.E.; Yun, S.; Vashisht, A.; Zhang, Q.; Park, J.W.; Corey, E.; et al. Systemic surfaceome profiling identifies target antigens for immune-based therapy in subtypes of advanced prostate cancer. Proc. Natl. Acad. Sci. USA 2018, 115, E4473–E4482. [Google Scholar] [CrossRef]
  39. Powell, E.; Shao, J.; Picon, H.M.; Bristow, C.; Ge, Z.; Peoples, M.; Robinson, F.; Jeter-Jones, S.L.; Schlosberg, C.; Grzeskowiak, C.L.; et al. A functional genomic screen in vivo identifies CEACAM5 as a clinically relevant driver of breast cancer metastasis. NPJ Breast Cancer 2018, 4, 9. [Google Scholar] [CrossRef]
  40. Detarya, M.; Sawanyawisuth, K.; Aphivatanasiri, C.; Chuangchaiya, S.; Saranaruk, P.; Sukprasert, L.; Silsirivanit, A.; Araki, N.; Wongkham, S.; Wongkham, C. The O-GalNAcylating enzyme GALNT5 mediates carcinogenesis and progression of cholangiocarcinoma via activation of AKT/ERK signaling. Glycobiology 2020, 30, 312–324. [Google Scholar] [CrossRef]
  41. Guo, H.; Zhao, L.; Shi, B.; Bao, J.; Zheng, D.; Zhou, B.; Shi, J. GALNT5 uaRNA promotes gastric cancer progression through its interaction with HSP90. Oncogene 2018, 37, 4505–4517. [Google Scholar] [CrossRef] [PubMed]
  42. Qiu, H.; Shen, X.; Chen, B.; Chen, T.; Feng, G.; Chen, S.; Feng, D.; Xu, Q. miR-30b-5p inhibits cancer progression and enhances cisplatin sensitivity in lung cancer through targeting LRP8. Apoptosis 2021, 26, 261–276. [Google Scholar] [CrossRef]
  43. Cai, J.; Chen, J.; Wu, T.; Cheng, Z.; Tian, Y.; Pu, C.; Shi, W.; Suo, X.; Wu, X.; Zhang, K. Genome-scale CRISPR activation screening identifies a role of LRP8 in Sorafenib resistance in Hepatocellular carcinoma. Biochem. Biophys. Res. Commun. 2020, 526, 1170–1176. [Google Scholar] [CrossRef] [PubMed]
  44. Li, L.; Qu, W.H.; Ma, H.P.; Wang, L.L.; Zhang, Y.B.; Ma, Y. LRP8, modulated by miR-1262, promotes tumour progression and forecasts the prognosis of patients in breast cancer. Arch. Physiol. Biochem. 2020, 128, 657–665. [Google Scholar] [CrossRef] [PubMed]
  45. Lin, C.C.; Lo, M.C.; Moody, R.; Jiang, H.; Harouaka, R.; Stevers, N.; Tinsley, S.; Gasparyan, M.; Wicha, M.; Sun, D. Targeting LRP8 inhibits breast cancer stem cells in triple-negative breast cancer. Cancer Lett. 2018, 438, 165–173. [Google Scholar] [CrossRef]
  46. Kim, S.; Kang, H.Y.; Nam, E.H.; Choi, M.S.; Zhao, X.F.; Hong, C.S.; Lee, J.W.; Lee, J.H.; Park, Y.K. TMPRSS4 induces invasion and epithelial-mesenchymal transition through upregulation of integrin alpha5 and its signaling pathways. Carcinogenesis 2010, 31, 597–606. [Google Scholar] [CrossRef]
  47. Panebianco, C.; Trivieri, N.; Villani, A.; Terracciano, F.; Latiano, T.P.; Potenza, A.; Perri, F.; Binda, E.; Pazienza, V. Improving Gemcitabine Sensitivity in Pancreatic Cancer Cells by Restoring miRNA-217 Levels. Biomolecules 2021, 11, 639. [Google Scholar] [CrossRef]
  48. Wang, F.; Xiang, Z.; Huang, T.; Zhang, M.; Zhou, W.B. ANLN Directly Interacts with RhoA to Promote Doxorubicin Resistance in Breast Cancer Cells. Cancer Manag. Res. 2020, 12, 9725–9734. [Google Scholar] [CrossRef]
  49. Moyano-Galceran, L.; Pietila, E.A.; Turunen, S.P.; Corvigno, S.; Hjerpe, E.; Bulanova, D.; Joneborg, U.; Alkasalias, T.; Miki, Y.; Yashiro, M.; et al. Adaptive RSK-EphA2-GPRC5A signaling switch triggers chemotherapy resistance in ovarian cancer. EMBO Mol. Med. 2020, 12, e11177. [Google Scholar] [CrossRef]
  50. Szklarczyk, D.; Gable, A.L.; Lyon, D.; Junge, A.; Wyder, S.; Huerta-Cepas, J.; Simonovic, M.; Doncheva, N.T.; Morris, J.H.; Bork, P.; et al. STRING v11: Protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 2019, 47, D607–D613. [Google Scholar] [CrossRef]
  51. Lever, J.; Zhao, E.Y.; Grewal, J.; Jones, M.R.; Jones, S.J.M. CancerMine: A literature-mined resource for drivers, oncogenes and tumor suppressors in cancer. Nat. Methods 2019, 16, 505–507. [Google Scholar] [CrossRef] [PubMed]
  52. Fabregat, A.; Sidiropoulos, K.; Garapati, P.; Gillespie, M.; Hausmann, K.; Haw, R.; Jassal, B.; Jupe, S.; Korninger, F.; McKay, S.; et al. The Reactome pathway Knowledgebase. Nucleic Acids Res. 2016, 44, D481–D487. [Google Scholar] [CrossRef] [PubMed]
  53. Peng, J.; Sun, B.F.; Chen, C.Y.; Zhou, J.Y.; Chen, Y.S.; Chen, H.; Liu, L.; Huang, D.; Jiang, J.; Cui, G.S.; et al. Single-cell RNA-seq highlights intra-tumoral heterogeneity and malignant progression in pancreatic ductal adenocarcinoma. Cell Res. 2019, 29, 725–738. [Google Scholar] [CrossRef]
  54. Butler, A.; Hoffman, P.; Smibert, P.; Papalexi, E.; Satija, R. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat. Biotechnol. 2018, 36, 411–420. [Google Scholar] [CrossRef]
  55. Zhang, X.; Lan, Y.; Xu, J.; Quan, F.; Zhao, E.; Deng, C.; Luo, T.; Xu, L.; Liao, G.; Yan, M.; et al. CellMarker: A manually curated resource of cell markers in human and mouse. Nucleic Acids Res. 2019, 47, D721–D728. [Google Scholar] [CrossRef] [PubMed]
  56. Iorio, F.; Knijnenburg, T.A.; Vis, D.J.; Bignell, G.R.; Menden, M.P.; Schubert, M.; Aben, N.; Goncalves, E.; Barthorpe, S.; Lightfoot, H.; et al. A Landscape of Pharmacogenomic Interactions in Cancer. Cell 2016, 166, 740–754. [Google Scholar] [CrossRef]
  57. Finan, C.; Gaulton, A.; Kruger, F.A.; Lumbers, R.T.; Shah, T.; Engmann, J.; Galver, L.; Kelley, R.; Karlsson, A.; Santos, R.; et al. The druggable genome and support for target identification and validation in drug development. Sci. Transl. Med. 2017, 9, eaag1166. [Google Scholar] [CrossRef]
  58. Ellrott, K.; Bailey, M.H.; Saksena, G.; Covington, K.R.; Kandoth, C.; Stewart, C.; Hess, J.; Ma, S.; Chiotti, K.E.; McLellan, M.; et al. Scalable Open Science Approach for Mutation Calling of Tumor Exomes Using Multiple Genomic Pipelines. Cell Syst. 2018, 6, 271–281.e7. [Google Scholar] [CrossRef]
  59. Newman, A.M.; Liu, C.L.; Green, M.R.; Gentles, A.J.; Feng, W.; Xu, Y.; Hoang, C.D.; Diehn, M.; Alizadeh, A.A. Robust enumeration of cell subsets from tissue expression profiles. Nat. Methods 2015, 12, 453–457. [Google Scholar] [CrossRef]
Figure 1. Identification of UDEGs in PDAC. (A) Workflow for identifying UDEGs in PDAC. (B) Overlapping genes in two DEG lists were defined as UDEGs. (C) Dysregulation frequencies of nineteen UDEGs. (D) Brief information on UDEGs.
Figure 1. Identification of UDEGs in PDAC. (A) Workflow for identifying UDEGs in PDAC. (B) Overlapping genes in two DEG lists were defined as UDEGs. (C) Dysregulation frequencies of nineteen UDEGs. (D) Brief information on UDEGs.
Ijms 26 03936 g001
Figure 2. Functional analysis of UDEGs. (A) UDEGs and their direct neighbors in PPI network. Light blue represents neighbor genes, red represents upregulated genes, and green represents downregulated genes. (B) KEGG enrichment analysis of UDEGs and their direct neighbor genes. (C) Pathways with different functions that are hierarchically enriched with UDEGs according to Reactome.
Figure 2. Functional analysis of UDEGs. (A) UDEGs and their direct neighbors in PPI network. Light blue represents neighbor genes, red represents upregulated genes, and green represents downregulated genes. (B) KEGG enrichment analysis of UDEGs and their direct neighbor genes. (C) Pathways with different functions that are hierarchically enriched with UDEGs according to Reactome.
Ijms 26 03936 g002
Figure 3. Single-cell analysis of 24 PDAC and 11 control samples. t-SNE plot of clustering and cell-type annotation of tumor (A) and normal (B) samples. Cell numbers for each cell type in tumor (C) and normal (D) samples. Expression of UDEGs in different cell types in tumor (E) and normal (F) samples.
Figure 3. Single-cell analysis of 24 PDAC and 11 control samples. t-SNE plot of clustering and cell-type annotation of tumor (A) and normal (B) samples. Cell numbers for each cell type in tumor (C) and normal (D) samples. Expression of UDEGs in different cell types in tumor (E) and normal (F) samples.
Ijms 26 03936 g003
Figure 4. Correlation analysis between expression of UDEGs and resistance to anticancer drugs. (A) Overview of correlations between IC50 of drug in pancreatic cells and UDEG expression. (B) Overview of significant correlations between drug resistance and expression of UDEGs. (C) Survival analysis of ANLN, GPRC5A and SERPINB5 in TCGA PDAC patients. (D) DGI network of UDEGs and their PPI partners. Light blue represents neighbor genes, red represents upregulated genes, green represents downregulated genes, and purple represents drugs.
Figure 4. Correlation analysis between expression of UDEGs and resistance to anticancer drugs. (A) Overview of correlations between IC50 of drug in pancreatic cells and UDEG expression. (B) Overview of significant correlations between drug resistance and expression of UDEGs. (C) Survival analysis of ANLN, GPRC5A and SERPINB5 in TCGA PDAC patients. (D) DGI network of UDEGs and their PPI partners. Light blue represents neighbor genes, red represents upregulated genes, green represents downregulated genes, and purple represents drugs.
Ijms 26 03936 g004
Figure 5. Roles of UDEGs in response of PDAC patients to immunotherapy. (A). Overview of immune cell proportions in TCGA PDAC patients. (B). Correlation between UDEGs and immune cell proportions. “×” represents a non-significant correlation. Genes labeled in green are down-regulated, and those in red are up-regulated.
Figure 5. Roles of UDEGs in response of PDAC patients to immunotherapy. (A). Overview of immune cell proportions in TCGA PDAC patients. (B). Correlation between UDEGs and immune cell proportions. “×” represents a non-significant correlation. Genes labeled in green are down-regulated, and those in red are up-regulated.
Ijms 26 03936 g005
Table 1. Functional enrichment analysis of different cell types in single-cell analysis of pancreatic cancer.
Table 1. Functional enrichment analysis of different cell types in single-cell analysis of pancreatic cancer.
Cell TypeNumber of PathwaysConsistance
NormalTumorCommon
Acinar cells56355.00%
Cancer-related ductal cells-29--
Ductal cells35302268.10%
Endocrine cells27292278.67%
Endothelial cells94826473.07%
Fibroblasts22291663.95%
Macrophages70746488.96%
Stellate cells39282370.56%
T cells30322890.42%
Table 2. Overview of samples used in this study.
Table 2. Overview of samples used in this study.
GEO AccessionPlatformSample Size
PDACPDAC_AdjacentNormal
Unpaired Samples
GSE62452GPL624469610
E-MTAB-1791A-MEXP-22712687441
GSE91035GPL227632508
GSE62165GPL13667118130
GSE56560GPL51752843
GSE71989GPL5701308
Total 521212
Paired Samples -
GSE60646GPL51751010-
GSE22780GPL57088-
GSE28735GPL62444545-
GSE15471GPL5703939-
Total 102102-
PDAC: pancreatic ductal adenocarcinoma.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Xia, J.; Zheng, L.; Zhang, H.; Fan, Q.; Liu, H.; Wang, O.; Yan, H. Drug Resistance Analysis of Pancreatic Cancer Based on Universally Differentially Expressed Genes. Int. J. Mol. Sci. 2025, 26, 3936. https://doi.org/10.3390/ijms26093936

AMA Style

Xia J, Zheng L, Zhang H, Fan Q, Liu H, Wang O, Yan H. Drug Resistance Analysis of Pancreatic Cancer Based on Universally Differentially Expressed Genes. International Journal of Molecular Sciences. 2025; 26(9):3936. https://doi.org/10.3390/ijms26093936

Chicago/Turabian Style

Xia, Jie, Linyong Zheng, Huarong Zhang, Qi Fan, Hui Liu, Ouxi Wang, and Haidan Yan. 2025. "Drug Resistance Analysis of Pancreatic Cancer Based on Universally Differentially Expressed Genes" International Journal of Molecular Sciences 26, no. 9: 3936. https://doi.org/10.3390/ijms26093936

APA Style

Xia, J., Zheng, L., Zhang, H., Fan, Q., Liu, H., Wang, O., & Yan, H. (2025). Drug Resistance Analysis of Pancreatic Cancer Based on Universally Differentially Expressed Genes. International Journal of Molecular Sciences, 26(9), 3936. https://doi.org/10.3390/ijms26093936

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop